51
|
Zhu N, Huang B, Zhu L. Bibliometric analysis of the inflammation in diabetic cardiomyopathy. Front Cardiovasc Med 2022; 9:1006213. [PMID: 36582738 PMCID: PMC9792483 DOI: 10.3389/fcvm.2022.1006213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Background Maladaptive inflammation is implicated in the development of diabetic cardiomyopathy (DCM). This study aimed to visually analyze the global scientific output over the past two decades regarding research on inflammation associated with DCM. Methods All relevant articles and reviews were retrieved in the Web of Science (WOS) Core Collection (limited to SCIE) using "inflammation" and "diabetic cardiomyopathy" as search terms. Articles and reviews published from 1 January 2001 to 28 February 2021 were collected. Visualization analysis and statistical analysis were conducted by Microsoft 365 Excel and VOSviewer 1.6.18. Results A total of 578 documents were finally selected for further analysis. The publications regarding inflammation and DCM increased gradually over approximately 20 years. The most prolific country was China, with 296 documents and the most citations (9,366). The most influential author groups were Lu Cai and Yihui Tan who were from the United States. The bibliometric analysis of co-occurrence keywords showed that inflammation in DCM is composed of numerous molecules (NF-κB, NLRP3 inflammasome, Nrf-2, TNF-α, protein kinase C, PPARα, TLR4, p38 mitogen-activated protein kinase, TGF-β, Sirt1, and AKT), a variety of cardiac cell types (stem cell, fibroblast, and cardiomyocyte), physiological processes (apoptosis, oxidative stress, autophagy, endoplasmic reticulum stress, hypertrophy, mitochondrion dysfunction, and proliferation), and drugs (sulforaphane, metformin, empagliflozin, and rosuvastatin). Conclusion Our bibliometric analysis presents the characteristics and trends of inflammation in DCM and shows that research on inflammation in DCM will continue to be a hotspot.
Collapse
Affiliation(s)
- Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, Wenzhou People’s Hospital, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China,*Correspondence: Ning Zhu,
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liuyan Zhu
- Department of General Practice, The Third Affiliated Hospital of Shanghai University, Wenzhou People’s Hospital, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
52
|
Qian Y, Yang Y, Qing W, Li C, Kong M, Kang Z, Zuo Y, Wu J, Yu M, Yang Z. Coxsackievirus B3 infection induces glycolysis to facilitate viral replication. Front Microbiol 2022; 13:962766. [PMID: 36569097 PMCID: PMC9780277 DOI: 10.3389/fmicb.2022.962766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is a leading cause of viral myocarditis, but no effective treatment strategy against CVB3 is available. Viruses lack an inherent metabolic system and thus depend on host cellular metabolism for their benefit. In this study, we observed that CVB3 enhanced glycolysis in H9c2 rat cardiomyocytes and HL-1 mouse cardiomyocytes. Therefore, three key glycolytic enzymes, namely, hexokinase 2 (HK2), muscle phosphofructokinase (PFKM), and pyruvate kinase M2 (PKM2), were measured in CVB3-infected H9c2 and HL-1 cells. Expression levels of HK2 and PFKM, but not PKM2, were increased in CVB3-infected H9c2 cells. All three key glycolytic enzymes showed elevated expression in CVB3-infected HL-1 cells. To further investigate this, we used 2 deoxyglucose, sodium citrate, and shikonin as glycolysis inhibitors for HK2, PFKM, and PKM2, respectively. Glycolysis inhibitors significantly reduced CVB3 replication, while the glycolysis enhancer dramatically promoted it. In addition, glycolysis inhibitors decreased autophagy and accelerated autophagosome degradation. The autophagy inducer eliminated partial inhibition effects of glycolysis inhibitors on CVB3 replication. These results demonstrate that CVB3 infection enhances glycolysis and thus benefits viral replication.
Collapse
Affiliation(s)
- Yujie Qian
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yeyi Yang
- Department of Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenxiang Qing
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chunyun Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Min Kong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhijuan Kang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuanbojiao Zuo
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiping Wu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Meng Yu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zuocheng Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Zuocheng Yang
| |
Collapse
|
53
|
Luo W, Wu G, Chen X, Zhang Q, Zou C, Wang J, Liu J, Chattipakorn N, Wang Y, Liang G. Blockage of MyD88 in cardiomyocytes alleviates cardiac inflammation and cardiomyopathy in experimental diabetic mice. Biochem Pharmacol 2022; 206:115292. [DOI: 10.1016/j.bcp.2022.115292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022]
|
54
|
Luo W, Lin K, Hua J, Han J, Zhang Q, Chen L, Khan ZA, Wu G, Wang Y, Liang G. Schisandrin B Attenuates Diabetic Cardiomyopathy by Targeting MyD88 and Inhibiting MyD88-Dependent Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202590. [PMID: 36180407 PMCID: PMC9631063 DOI: 10.1002/advs.202202590] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/19/2022] [Indexed: 06/16/2023]
Abstract
Diabetes manifests as chronic inflammation and leads to the development diabetic cardiomyopathy (DCM). Targeting key proteins in inflammatory signaling may provide new therapy for DCM. In this study, the authors explore the pharmacological effects and mechanisms of Schisandrin B (Sch B), a natural compound with anti-inflammatory activity against DCM. It is shown that Sch B prevents high-level glucose (HG)-induced hypertrophic and fibrotic responses in cultured cardiomyocytes. RNA sequencing and inflammatory qPCR microarray show that Sch B mainly affects myeloid differentiation primary response 88 (MyD88)-dependent inflammatory gene expression in HG-challenged cardiomyocytes. Further studies indicate that Sch B directly binds to and inhibits MyD88 activation, but does not alter MyD88-independent Toll-like receptor signaling in vivo and in vitro. Inhibiting or silencing MyD88 is associated with reduced levels of HG-induced inflammatory cytokines and myocardial injuries in vitro. Treatment of type 1 and type 2 diabetic mice with Sch B protects heart function, reduces myocardial injuries, and decreases secretion of inflammatory cytokines. Cardiomyocyte-specific MyD88 knockout also protects mice against cardiac inflammation and injury in type 1 diabetic mice. In conclusion, these studies show that cardiomyocyte MyD88 plays an apathogenetic role in DCM and Sch B specifically targets MyD88 to reduce inflammatory DCM.
Collapse
Affiliation(s)
- Wu Luo
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiang311399China
- Department of Cardiology and Medical Research Centerthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Ke Lin
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
- Department of Cardiology and Medical Research Centerthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Junyi Hua
- Department of Cardiovascular Medicinethe Second Affiliated Hospital of Zhejiang University of Traditional Chinese MedicineHangzhouZhejiang310009China
- Department of Cardiovascular MedicineQuzhou Hospital of Traditional Chinese Medicine (Four Provincial Marginal Hospitals of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine)QuzhouZhejiang324002China
| | - Jibo Han
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Qiuyan Zhang
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Lingfeng Chen
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiang311399China
| | - Zia A. Khan
- Department of Pathology and Laboratory MedicineUniversity of Western OntarioLondonOntarioN6A 5C1Canada
| | - Gaojun Wu
- Department of Cardiology and Medical Research Centerthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Yi Wang
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
| | - Guang Liang
- Chemical Biology Research CenterSchool of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiang325035China
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiang311399China
- Department of Cardiology and Medical Research Centerthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325035China
| |
Collapse
|
55
|
Ye B, Shi X, Xu J, Dai S, Xu J, Fan X, Han B, Han J. Gasdermin D mediates doxorubicin-induced cardiomyocyte pyroptosis and cardiotoxicity via directly binding to doxorubicin and changes in mitochondrial damage. Transl Res 2022; 248:36-50. [PMID: 35545198 DOI: 10.1016/j.trsl.2022.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/25/2022] [Accepted: 05/03/2022] [Indexed: 12/21/2022]
Abstract
Doxorubicin (Dox), as a widely used anthracycline antitumor drug, can cause severe cardiotoxicity. Cardiomyocyte death and inflammation are involved in the pathophysiology of Dox-induced cardiotoxicity (DIC). Gasdermin D (GSDMD) is known as a key executioner of pyroptosis, which is a pro-inflammatory programmed cell death. We aimed to investigate the impact of GSDMD on DIC and systematically reveal its underlying mechanisms. Our findings indicated that Dox induced cardiomyocyte pyroptosis in a GSDMD-dependent manner by utilizing siRNA or overexpression-plasmid technique. We then generated GSDMD global knockout mice via CRISPR/Cas9 system and found that GSDMD deficiency reduced Dox-induced cardiomyopathy. Dox induced the activation of inflammatory caspases, which subsequently mediated GSDMD-N generation indirectly. Using molecular dynamics simulation and cell-free systems, we confirmed that Dox directly bound to GSDMD and facilitated GSDMD-N-mediated pyroptosis. Furthermore, GSDMD also mediated Dox-induced mitochondrial damage via Bnip3 and mitochondrial perforation in cardiomyocytes. These findings provide fresh insights into the mechanism of how Dox-engaged GSDMD orchestrates adverse cardiotoxicity and highlight the prospects of GSDMD as a potential target for DIC.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, Zhejiang 325000, China
| | - Xiaowen Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China
| | - Jianjiang Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China
| | - Shanshan Dai
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, Zhejiang 325000, China
| | - Jiajun Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China
| | - Xiaoxi Fan
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical University, WenZhou, Zhejiang 325000, China
| | - Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, China.
| |
Collapse
|
56
|
Chen J, Peng H, Chen C, Wang Y, Sang T, Cai Z, Zhao Q, Chen S, Lin X, Eling T, Wang X. NAG-1/GDF15 inhibits diabetic nephropathy via inhibiting AGE/RAGE-mediated inflammation signaling pathways in C57BL/6 mice and HK-2 cells. Life Sci 2022; 311:121142. [DOI: 10.1016/j.lfs.2022.121142] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
|
57
|
Lin J, Lin K, Huang L, Jiang Y, Ding X, Luo W, Samorodov AV, Pavlov VN, Liang G, Qian J, Wang Y. Heme induces inflammatory injury by directly binding to the complex of myeloid differentiation protein 2 and toll-like receptor 4. Toxicol Lett 2022; 370:15-23. [PMID: 36115635 DOI: 10.1016/j.toxlet.2022.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/20/2022] [Accepted: 09/12/2022] [Indexed: 10/31/2022]
Abstract
Heme, as an essential component of hemoproteins, is a prosthetic co-factor found in many cells, which is essential for physiologically vital oxygen transport. However, extracellular or circulatory heme is cytotoxic and triggers inflammation. Although the proinflammatory role of heme has been reported to be associated with Toll-like receptor 4 (TLR4) signaling, the exact mechanism remains unknown. Here, we show that heme promotes TLR4 signaling and inflammation via directly physically interacting with TLR4 and its adaptor protein myeloid differentiation protein 2 (MD2). Genetic loss of MD2 ameliorates heme-induced inflammation and inflammatory cytokine production in the spleen of MD2 knockout (MD2-/-) mice. Using mouse macrophage RAW 264.7 cell line, we show that heme induces TLR4 dimerization and MD2/TLR4/MyD88 activation by physically interacting with TLR4 and MD2 in vitro. Genetic loss of MD2 inhibits heme-induced inflammation and MAPK/NF-κB pathway in mouse primary macrophages extracted from MD2-/- mice. Furthermore, pharmacological inhibition of MD2 by L6H9 ameliorates heme-induced inflammation in macrophages. These findings demonstrate that heme causes inflammation by directly binding to MD2/TLR4 complex, leading to activation of TLR4/MAPK/NF-κB signaling pathway and production of downstream effectors of inflammation.
Collapse
Affiliation(s)
- Jianjun Lin
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lijiang Huang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Yongsheng Jiang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Xiaoxiao Ding
- Department of pharmacy, the People' s Hospital of Beilun District , Ningbo, Zhejiang 315807, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Aleksandr V Samorodov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Valentin N Pavlov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yi Wang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
58
|
Zhan X, Cheng L, Huo N, Yu L, Liu C, Liu T, Li G, Fu H. Sodium-glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. Front Cardiovasc Med 2022; 9:908037. [PMID: 36148071 PMCID: PMC9485554 DOI: 10.3389/fcvm.2022.908037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The mechanism of sodium-glucose cotransporter-2 inhibitor (SGLT-2i) reducing the incidence of atrial fibrillation remains unclear. We hypothesize that sodium-glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. METHODS A total of 42 rats were randomly assigned into three groups: control group (CON group); diabetes group (DM group): diabetes mellitus rats were established by 65 mg/kg streptozotocin (STZ) intraperitoneal injection; and diabetes + dapagliflozin group (DM + DAPA group): diabetic rats were given DAPA gavage administration (DAPA 2mg/kg/d for 4 weeks by gavage administration), 14 rats in each group. Epicardial multiple-lead recording and intracardiac electrophysiology studies were performed to investigate the electrical remodeling in the heart and the atrial fibrillation inducibility in each group. Western blot analysis and real-time PCR were used to determine the protein and mRNA expression of toll-like receptor 4 (TLR4), interleukin receptor-associated kinase 1 (IRAK1), tumor necrosis factor receptor-associated factor 6 (TRAF6), nuclear factor-kappa B (NF-κB), and type I collagen (collagen I). RESULTS Compared with rats in CON group, rats in DM group showed marked myocardial fibrosis, ectopic pacing excitement, reduced conduction velocity, decreased cardiac function. TLR4/IRAK1/TRAF6/NF-κB, collagen I proteins expressions and incidence of atrial fibrillation (27.3%) were increased in DM group. Parts of these changes were reversed by treatment of DAPA. Incidence of atrial fibrillation was decreased in DM + DAPA group (2.8%). CONCLUSIONS SGLT-2i dapagliflozin may prevent diabetic rats' atrial remodeling and reduce the inducibility of atrial fibrillation partly by targeting TLR4/IRAK1/TRAF6/NF-κB inflammatory pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
59
|
Fan Z, Ma H, Li Y, Wu Y, Wang J, Xiong L, Fang Z, Zhang X. Neuronal MD2 induces long-term mental impairments in septic mice by facilitating necroptosis and apoptosis. Front Pharmacol 2022; 13:884821. [PMID: 36016572 PMCID: PMC9396348 DOI: 10.3389/fphar.2022.884821] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a complication of sepsis with high morbidity rates. Long-lasting mental health issues in patients with SAE result in a substantial decrease in quality of life. However, its underlying mechanism is unclear, and effective treatments are not available. In the current study, we explored the role of apoptosis and necroptosis related to mental dysfunction in sepsis. In a mouse model of sepsis constructed by cecal ligation and puncture (CLP), altered behavior was detected by the open field, elevated-plus maze and forced swimming tests on the fourteenth day. Moreover, apoptosis- and necroptosis-associated proteins and morphological changes were examined in the hippocampus of septic mice. Long-lasting depression-like behaviors were detected in the CLP mice, as well as significant increases in neuronal apoptosis and necroptosis. Importantly, we found that apoptosis and necroptosis were related according to Ramsay’s rule in the brains of the septic mice. Inhibiting myeloid differentiation factor 2 (MD2), the crosstalk mediator of apoptosis and necroptosis, in neurons effectively reduced neuronal loss and alleviated depression-like behaviors in the septic mice. These results suggest that neuronal death in the hippocampus contributes to the mental impairments in SAE and that inhibiting neuronal MD2 is a new strategy for treating mental health issues in sepsis by inhibiting necroptosis and apoptosis.
Collapse
Affiliation(s)
- Zhongmin Fan
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hongwei Ma
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yi Li
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - You Wu
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiajia Wang
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lize Xiong
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Zongping Fang
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Zongping Fang, ; Xijing Zhang,
| | - Xijing Zhang
- Department of Critical Care Medicine and Department of Anesthesiology and Perioprative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Zongping Fang, ; Xijing Zhang,
| |
Collapse
|
60
|
Zhu J, Zhang Y, Shi L, Xia Y, Zha H, Li H, Song Z. RP105 protects against ischemic and septic acute kidney injury via suppressing TLR4/NF-κB signaling pathways. Int Immunopharmacol 2022; 109:108904. [PMID: 35696803 DOI: 10.1016/j.intimp.2022.108904] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 12/31/2022]
Abstract
Acute kidney injury (AKI) is a critical and severe clinical disease caused by a variety of factors. Toll-like receptors (TLRs) play a crucial role in pathogenesis of AKI. Radioprotective 105 kDa protein (RP105) is a member of the TLR family, but the role of RP105 in AKI is unknown. In this study, we overexpressed RP105 in renal tissue and cultured proximal tubular cells in which we then induced ischemic and septic AKI. Renal structure injuries were examined by hematoxylin eosin staining, while renal function was assessed by measuring serum blood urea nitrogen (BUN) and creatinine (SCr) levels. The TUNEL assay was used to detect apoptosis induced changes in the expression of RP105, and nuclear factor κB (NF-κB) in renal tissue detected by Western blot. Inflammatory cytokines including iNOS, IL-1β, IL-6, and TNF-α were detected by quantitative real-time PCR. The inflammatory indicators, F4/80 and MPO, were identified by IHC staining. The results showed that expression of the TLR4/NF-kB signaling pathway was enhanced in renal ischemia-reperfusion injury and septic renal injury, and that overexpression of RP105 in renal tissue alleviated ischemic and septic AKI. Moreover, RP105 gene delivery was associated with reduced renal inflammatory cells infiltration and inflammatory cytokines after AKI. RP105 overexpression also inhibited nuclear translocation of NF-κB after AKI in both in vitro and in vivo, and blunted the interaction between Myeloid Differentiation factor 2 (MD2) and TLR4. These results indicated that RP105 protected against renal ischemic and septic AKI injury by suppressing inflammatory responses mediated by TLR4 signaling pathways. This study suggests that the anti-inflammatory roles of RP105 have potential for preventing and treating renal ischemic and septic AKI.
Collapse
Affiliation(s)
- Jiefu Zhu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yafei Zhang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China
| | - Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yao Xia
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China
| | - Huimin Li
- Department of Nephrology, Affiliated Renhe Hospital of Three Gorges University, Yichang, Hubei 443000, China
| | - Zhixia Song
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China.
| |
Collapse
|
61
|
Dai C, Luo W, Chen Y, Shen S, Wang Z, Chen R, Wang J, Chattipakorn N, Huang W, Liang G. Tabersonine attenuates Angiotensin II-induced cardiac remodeling and dysfunction through targeting TAK1 and inhibiting TAK1-mediated cardiac inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154238. [PMID: 35696800 DOI: 10.1016/j.phymed.2022.154238] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Angiotensin II (Ang II)-induced cardiac inflammation contribute to pathological cardiac remodeling and hypertensive heart failure (HF). Tabersonine (Tab) is an indole alkaloid mainly isolated from Catharanthus roseus and exhibits anti-inflammatory activity in various systems. However, the role of Tab in hypertensive HF and its molecular targets remains unknown. HYPOTHESIS/PURPOSE We aimed to investigate potential cardioprotective effects and mechanism of Tab against Ang II-induced cardiac injuries. METHODS C57BL/6 mice were administered Ang II (at 1000 ng/kg/min) by micro-osmotic pump infusion for 30 days to develop hypertensive HF. Tab at 20 and 40 mg/kg/day was administered during the last 2 weeks to elucidate the cardioprotective properties. Cultured cardiomyocyte-like H9c2 cells and rat primary cardiomyocytes were used for mechanistic studies of Tab. RESULTS We demonstrate for the first time that Tab provides protection against Ang II-induced cardiac dysfunction in mice, associated with reduced cardiac inflammation and fibrosis. Mechanistically, we show that Tab may interacts with TAK1 to inhibit Ang II-induced TAK1 ubiquitination and phosphorylation. Disruption of TAK1 activation by Tab blocked downstream NF-κB and JNK/P38 MAPK signaling activation and decreased cardiac inflammation and fibrosis both in vitro and in vivo. TAK1 knockdown also blocked Ang II-induced cardiomyocytes injuries and prevented the innately pharmacological effects of Tab. CONCLUSION Our results indicate that Tab protects hearts against Ang II-mediated injuries through targeting TAK1 and inhibiting TAK1-mediated inflammatory cascade and response. Thus, Tab may be a potential therapeutic candidate for hypertensive HF.
Collapse
Affiliation(s)
- Chengyi Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yanghao Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Siyuan Shen
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhe Wang
- Department of Pharmacy, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ruijie Chen
- Department of Pharmacy, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jun Wang
- Department of Cardiology, Wenzhou Central Hospital and Affiliated Dingli Clinical Institute, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Weijian Huang
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
62
|
Flavokawain B alleviates LPS-induced acute lung injury via targeting myeloid differentiation factor 2. Acta Pharmacol Sin 2022; 43:1758-1768. [PMID: 34737421 PMCID: PMC9253132 DOI: 10.1038/s41401-021-00792-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/08/2021] [Indexed: 12/16/2022]
Abstract
Acute lung injury (ALI) is a sudden onset systemic inflammatory response. ALI causes severe morbidity and death and currently no effective pharmacological therapies exist. Natural products represent an excellent resource for discovering new drugs. Screening anti-inflammatory compounds from the natural product bank may offer viable candidates for molecular-based therapies for ALI. In this study, 165 natural compounds were screened for anti-inflammatory activity in lipopolysaccharide (LPS)-challenged macrophages. Among the screened compounds, flavokawain B (FKB) significantly reduced LPS-induced pro-inflammatory IL-6 secretion in macrophages. FKB also reduced the formation of LPS/TLR4/MD2 complex by competitively binding to MD2, suppressing downstream MAPK and NF-κB signaling activation. Finally, FKB treatment of mice reduced LPS-induced lung injury, systemic and local inflammatory cytokine production, and macrophage infiltration in lungs. These protective activities manifested as increased survival in the ALI model, and reduced mortality upon bacterial infection. In summary, we demonstrate that the natural product FKB protects against LPS-induced lung injury and sepsis by interacting with MD2 and inhibiting inflammatory responses. FKB may potentially serve as a therapeutic option for the treatment of ALI.
Collapse
|
63
|
Cross-Talk between the Cytokine IL-37 and Thyroid Hormones in Modulating Chronic Inflammation Associated with Target Organ Damage in Age-Related Metabolic and Vascular Conditions. Int J Mol Sci 2022; 23:ijms23126456. [PMID: 35742902 PMCID: PMC9224418 DOI: 10.3390/ijms23126456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic inflammation is considered to be the main mechanism contributing to the development of age-related metabolic and vascular conditions. The phases of chronic inflammation that mediate the progression of target organ damage in these conditions are poorly known, however. In particular, there is a paucity of data on the link between chronic inflammation and metabolic disorders. Based on some of our own results and recent developments in our understanding of age-related inflammation as a whole-body response, we discuss the hypothesis that cross-talk between the cytokine IL-37 and thyroid hormones could be the key regulatory mechanism that justifies the metabolic effects of chronic tissue-related inflammation. The cytokine IL-37 is emerging as a strong natural suppressor of the chronic innate immune response. The effect of this cytokine has been identified in reversing metabolic costs of chronic inflammation. Thyroid hormones are known to regulate energy metabolism. There is a close link between thyroid function and inflammation in elderly individuals. Nonlinear associations between IL-37 and thyroid hormones, considered within the wider clinical context, can improve our understanding of the phases of chronic inflammation that are associated with target organ damage in age-related metabolic and vascular conditions.
Collapse
|
64
|
Nie K, Li J, Peng L, Zhang M, Huang W. Pan-Cancer Analysis of the Characteristics of LY96 in Prognosis and Immunotherapy Across Human Cancer. Front Mol Biosci 2022; 9:837393. [PMID: 35647025 PMCID: PMC9130738 DOI: 10.3389/fmolb.2022.837393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/25/2022] [Indexed: 12/28/2022] Open
Abstract
Lymphocyte antigen 96 (LY96) is implicated in tumorigenesis by modulating host immunity. However, an integrated pan-cancer analysis of LY96 in prognosis and immunotherapy across human cancers is still lacking. Therefore, we analyzed the LY96 expression and its prognostic role in tumors by multiple databases. We also investigated the correlation between LY96 and copy number, DNA methylation, somatic mutation, microsatellite instability (MSI), tumor mutation burden (TMB), tumor microenvironment (TME), and immune cell infiltration across human cancers. In addition, the biological processes related to LY96 across various tumors and the correlation between LY96 and 50% inhibitive concentration (IC50) of various drugs were investigated. We found that LY96 was differently expressed between tumor and normal tissues and was significantly upregulated in most types of cancers. LY96 was gradually upregulated from stages I to IV in several cancers. Moreover, we found LY96 may play a prognostic role in most cancers, and patients with high or low LY96 expression often show different clinical outcomes. LY96 was also associated with copy number, DNA methylation, somatic mutation, MSI, TMB, TME characteristics, and immune cell infiltration in cancers. LY96 may also regulate classic tumor-associated pathways in several cancers and is related to drug resistance. This article may help to elucidate the role of LY96 in tumorigenesis, which may promote the development of immunotherapy and targeted therapy in cancers.
Collapse
Affiliation(s)
- Kechao Nie
- Department of Integrated Traditional Chinese and Western Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Integrated Traditional Chinese and Western Internal Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Luqi Peng
- Department of Integrated Traditional Chinese and Western Internal Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Mei Zhang
- Department of Integrated Traditional Chinese and Western Internal Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Internal Medicine, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wei Huang,
| |
Collapse
|
65
|
Jia D, He Y, Wang Y, Xue M, Zhu L, Xia FX, Li Y, Gao Y, Li L, Chen S, Xu G, Yuan C. NEAT1: A novel long non-coding RNA involved in mediating type 2 diabetes and its various complications. Curr Pharm Des 2022; 28:1342-1350. [DOI: 10.2174/1381612828666220428093207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/03/2022] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Background:
Nuclear‐enriched abundant transcript 1 (abbreviated as NEAT1) refers to a long-chain non-coding RNA involved within various physiological and pathological processes. This study aimed at clarifying the effect and molecule system of neat1 within nonalcoholic fatty liver disease (NAFLD) as well as type 2 diabetes (T2DM).
Method:
In this review, we summarize and analyze current studies concerning mechanisms of NEAT1 in the development of type 2 diabetes and its complications. Also, we search the papers of NEAT1 in applying to NAFLD. The related studies were obtained through a systematic search of Pubmed.
Results:
Neat1 displays a close correlation with how T2DM occurs and develops, and it was confirmed to be significantly up-regulated in T2DM and its various complications (e.g., diabetics nephropathy, diabetics cardiomyopathy, diabetics retinopathy as well as diabetic neuropathy). Besides, neat1 is capable of impacting the occurrence, development and prognosis of NAFLD and T2DM.
Conclusion:
LncRNA neat1 is likely to act as a novel therapeutic target for and T2DM and its complications. Moreover, nonalcoholic fatty liver disease is also correlated with NEAT1.
Collapse
Affiliation(s)
- Dengke Jia
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yaping He
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yaqi Wang
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Mengzhen Xue
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Leiqi Zhu
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Fangqi Xia Xia
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yuanyang Li
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yan Gao
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Luoying Li
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Silong Chen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Guangfu Xu
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges, Yichang. Hubei 443002. China
| |
Collapse
|
66
|
Zhu N, Zhu L, Huang B, Xiang W, Zhao X. Galectin-3 Inhibition Ameliorates Streptozotocin-Induced Diabetic Cardiomyopathy in Mice. Front Cardiovasc Med 2022; 9:868372. [PMID: 35557520 PMCID: PMC9086782 DOI: 10.3389/fcvm.2022.868372] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Diabetic cardiomyopathy (DCM), characterized by cardiomyopathy with the absence of coronary artery disease, hypertension, and valvular heart disease in patients with diabetes, significantly increases the risk of heart failure. Galectin-3 (Gal-3) has been shown to regulate cardiac inflammation and fibrosis, but its role in DCM remains unclear. This study aimed to determine whether Gal-3 inhibition attenuates DCM and NF-κB p65 activation. Methods Diabetic cardiomyopathy (DCM) was established by intraperitoneal (IP) injection of streptozotocin for 5 consecutive days in mice. Myocardial injury markers, such as creatine kinase isoenzyme (CK-BM) and lactate dehydrogenase, were detected using ELISA. We used non-invasive transthoracic echocardiography to examine cardiac structure and function. Histological staining was used to explore myocardial morphology and fibrosis. Profibrotic markers and inflammatory cytokines were detected by ELISA and real-time PCR in vivo. The terminal deoxyribonucleotide transferasemediated dUTP nick end-labeling (TUNEL) and immunofluorescence assays were conducted to examine myocardial apoptosis and oxidative stress. Inflammatory cytokines induced by high glucose (HG) were also found in RAW264.7 macrophages. The underlying molecular mechanisms were determined using immunofluorescence and Western blotting analyses. Results The Gal-3 knockdown was observed to ameliorate myocardial apoptosis, oxidative stress, inflammatory cytokines release, macrophage infiltration, and fibrosis, thus, decreasing cardiac dysfunction in DCM mice. In addition, the silence of Gal-3 could suppress macrophage infiltration and inflammatory cytokine release induced by HG. Finally, a Gal-3/NF-κB p65 regulatory network was clarified in the pathogenesis of DCM. Conclusion The Gal-3 may promote myocardial apoptosis, oxidative stress, inflammation, and fibrosis in vivo and in vitro by the mechanism of reduction of NF-κB p65 activation.
Collapse
Affiliation(s)
- Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China,*Correspondence: Ning Zhu
| | - Liuyan Zhu
- Department of General Practice, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjun Xiang
- Department of Pathology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Xuyong Zhao
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| |
Collapse
|
67
|
Plin5, a New Target in Diabetic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2122856. [PMID: 35509833 PMCID: PMC9060988 DOI: 10.1155/2022/2122856] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
Abnormal lipid accumulation is commonly observed in diabetic cardiomyopathy (DC), which can create a lipotoxic microenvironment and damage cardiomyocytes. Lipid toxicity is an important pathogenic factor due to abnormal lipid accumulation in DC. As a lipid droplet (LD) decomposition barrier, Plin5 can protect LDs from lipase decomposition and regulate lipid metabolism, which is involved in the occurrence and development of cardiovascular diseases. In recent years, studies have shown that Plin5 expression is involved in the pathogenesis of DC lipid toxicity, such as oxidative stress, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and insulin resistance (IR) and has become a key target of DC research. Therefore, understanding the relationship between Plin5 and DC progression as well as the mechanism of this process is crucial for developing new therapeutic approaches and exploring new therapeutic targets. This review is aimed at exploring the latest findings and roles of Plin5 in lipid metabolism and DC-related pathogenesis, to explore possible clinical intervention approaches.
Collapse
|
68
|
Han X, Zhang J, Zhou L, Wei J, Tu Y, Shi Q, Zhang Y, Ren J, Wang Y, Ying H, Liang G. Sclareol ameliorates hyperglycemia‐induced renal injury through inhibiting the
MAPK
/
NF‐κB
signaling pathway. Phytother Res 2022; 36:2511-2523. [DOI: 10.1002/ptr.7465] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/26/2022] [Accepted: 04/01/2022] [Indexed: 12/29/2022]
Affiliation(s)
- Xue Han
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research Hangzhou Medical College Hangzhou China
- School of Pharmaceutical Sciences Hangzhou Medical College Hangzhou China
| | - Jiajia Zhang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research Hangzhou Medical College Hangzhou China
| | - Li Zhou
- College of Pharmaceutical Science Zhejiang Chinese Medical University Hangzhou China
| | - Jiajia Wei
- School of Pharmaceutical Sciences Hangzhou Medical College Hangzhou China
| | - Yu Tu
- School of Pharmaceutical Sciences Hangzhou Medical College Hangzhou China
| | - Qiaojuan Shi
- School of Pharmaceutical Sciences Hangzhou Medical College Hangzhou China
| | - Yi Zhang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research Hangzhou Medical College Hangzhou China
| | - Juan Ren
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research Hangzhou Medical College Hangzhou China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| | - Huazhong Ying
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research Hangzhou Medical College Hangzhou China
- College of Pharmaceutical Science Zhejiang Chinese Medical University Hangzhou China
| | - Guang Liang
- School of Pharmaceutical Sciences Hangzhou Medical College Hangzhou China
- Chemical Biology Research Center, School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| |
Collapse
|
69
|
Liu H, Lan W. Alleviation of Myocardial Inflammation in Diabetic Rats by Flavonoid Extract of Helichrysum Arenarium and Its Effect on Damaged Myocardial Cells Induced by High Glucose. Front Surg 2022; 9:873010. [PMID: 35495751 PMCID: PMC9046775 DOI: 10.3389/fsurg.2022.873010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 11/23/2022] Open
Abstract
Objective To investigate the effects of helichrysum arenarium flavonoid extract on high glucose damaged cardiomyocytes and the alleviation of myocardial inflammation in diabetic rats. Methods The study was divided into two parts, the first part was a cellular experiment in which a high-glucose cardiomyocyte injury model (H9C2) was established using a high-glucose culture medium, divided into low (group N1, 6.25 μg/mL), medium (group N2, 12.5 μg/mL), high dose group (group N3, 25 μg/mL) of helichrysum arenarium intervention and a model control group. The levels of enzyme activities [creatine kinase (CK) and lactate dehydrogenase (LDH)] in each group of H9c2 cells were measured by Enzyme-linked immunosorbent assay (ELISA), the expression levels of apoptotic proteins (Bax and Bcl-2) by western blot (WB), and the expression levels of inflammatory factors [tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6)] by RT-qPCR. The second part is animal experiments, after establishing the diabetic rat model, we used helichrysum arenarium flavonoid extract to intervene SD rats, divided into helichrysum arenarium intervention low (group S1, 250 mg/kg), medium (group S2, 500 mg/kg), high dose group (group S3, 1 g/kg), SD rat model group. Hematoxylin-eosin (HE) staining was used to observe myocardial tissue lesions, and Real Time Quantitative PCR (RT-qPCR) method was used to detect inflammatory (TNF-α, IL-1β, and IL-6) infiltration in myocardial tissue. Results Cellular experiments: The activity levels of enzymes such as CK and LDH and the levels of inflammatory factors such as TNF-α, IL-1β, and IL-6 in damaged cardiac myocytes were significantly decreased after helichrysum arenarium intervention; the expression levels of Bax protein were significantly down-regulated and the expression levels of Bcl-2 protein expression were significantly up-regulated. Animal experiment: HE staining showed that the model group had widened intercellular spaces, interstitial edema and obvious inflammatory cell infiltration in cardiac muscle tissue. After the intervention of helichrysum arenarium, the collagen fibers of rat myocardial cells were significantly reduced and cell degeneration was alleviated. Animal experiment: HE staining showed that the model group had widened intercellular spaces, interstitial edema and obvious inflammatory cell infiltration in cardiac muscle tissue. After the intervention of helichrysum arenarium, the collagen fibers of rat myocardial cells were significantly reduced and cell degeneration was alleviated; the levels of TNF-α, IL-1β, IL-6 and other inflammatory factors in myocardial tissues were significantly decreased. Conclusion The helichrysum arenarium flavonoid extract can reduce the degree of damage of H9C2 cells induced by high glucose and decrease the cellular inflammatory response, and its mechanism of action may be achieved by regulating the apoptotic factors Bax and Bcl-2. In addition, the extract of helichrysum arenarium can reduce the histopathological damage of myocardium in diabetic rats, decrease the inflammatory response in the tissue, and achieve the effect of myocardial protection.
Collapse
|
70
|
Phang RJ, Ritchie RH, Hausenloy DJ, Lees JG, Lim SY. Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy. Cardiovasc Res 2022; 119:668-690. [PMID: 35388880 PMCID: PMC10153440 DOI: 10.1093/cvr/cvac049] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/16/2022] [Accepted: 03/05/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with Type 2 diabetes mellitus (T2DM) frequently exhibit a distinctive cardiac phenotype known as diabetic cardiomyopathy. Cardiac complications associated with T2DM include cardiac inflammation, hypertrophy, fibrosis and diastolic dysfunction in the early stages of the disease, which can progress to systolic dysfunction and heart failure. Effective therapeutic options for diabetic cardiomyopathy are limited and often have conflicting results. The lack of effective treatments for diabetic cardiomyopathy is due in part, to our poor understanding of the disease development and progression, as well as a lack of robust and valid preclinical human models that can accurately recapitulate the pathophysiology of the human heart. In addition to cardiomyocytes, the heart contains a heterogeneous population of non-myocytes including fibroblasts, vascular cells, autonomic neurons and immune cells. These cardiac non-myocytes play important roles in cardiac homeostasis and disease, yet the effect of hyperglycaemia and hyperlipidaemia on these cell types are often overlooked in preclinical models of diabetic cardiomyopathy. The advent of human induced pluripotent stem cells provides a new paradigm in which to model diabetic cardiomyopathy as they can be differentiated into all cell types in the human heart. This review will discuss the roles of cardiac non-myocytes and their dynamic intercellular interactions in the pathogenesis of diabetic cardiomyopathy. We will also discuss the use of sodium-glucose cotransporter 2 inhibitors as a therapy for diabetic cardiomyopathy and their known impacts on non-myocytes. These developments will no doubt facilitate the discovery of novel treatment targets for preventing the onset and progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rebecca H Ritchie
- School of Biosciences, Parkville, Victoria 3010, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung City, Taiwan
| | - Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia.,Departments of Surgery and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
71
|
Lei J, Xiang P, Zeng S, Chen L, Zhang L, Yuan Z, Zhang J, Wang T, Yu R, Zhang W, Ibrahim II, Ma L, Yu C. Tetramethylpyrazine Alleviates Endothelial Glycocalyx Degradation and Promotes Glycocalyx Restoration via TLR4/NF-κB/HPSE1 Signaling Pathway During Inflammation. Front Pharmacol 2022; 12:791841. [PMID: 35185540 PMCID: PMC8850260 DOI: 10.3389/fphar.2021.791841] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Tetramethylpyrazine (TMP), a Chinese traditional herbal extraction widely used in treating cardiovascular diseases, could attenuate vascular endothelial injuries, but the underlying mechanism remains incomprehensive. Vascular glycocalyx coating on the endothelium would be damaged and caused endothelial dysfunction in the inflammatory microenvironment, which was the initial factor of morbidity of many vascular diseases, such as atherosclerosis (AS). Here, we thoroughly investigated the molecular mechanism of TMP on vascular endothelial glycocalyx in the LPS-induced inflammatory model both in vitro and in vivo. Results showed that pretreatment with TMP significantly inhibited glycocalyx degradation and monocytes adhesion to the endothelial process. Moreover, TMP pretreatment inhibited the expression of HPSE1 (a major degrading enzyme of endothelial glycocalyx), Toll-like receptor 4 (TLR4), and the translocation of nuclear factor kappa B p65 (NF-κB p65). We were utilized withTLR4 siRNA, NF-κB inhibitor, and HPSE1 overexpression analysis confirmed TMP's protection on endothelial glycocalyx injury, which further contributed to the monocyte-endothelial adhesion process. It was indicated that TMP might suppress glycocalyx degradation through TLR4/NF-κB/HPSE1 signaling pathway. Taken together, our results enriched the occurrence molecular mechanism of glycocalyx shedding and molecular regulation mechanism of TMP in protecting integrity of the glycocalyx structure during inflammation. As TMP is currently used in clinical applications, it may be considered a novel strategy against atherosclerosis through its ability to protect endothelial glycocalyx.
Collapse
Affiliation(s)
- Jin Lei
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Peng Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Shengmei Zeng
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Le Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Lei Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Jun Zhang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Institute of Life Sciences, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Ruihong Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Issa Issoufou Ibrahim
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| |
Collapse
|
72
|
Advanced Glycation End Products: A Sweet Flavor That Embitters Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms23052404. [PMID: 35269546 PMCID: PMC8910157 DOI: 10.3390/ijms23052404] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies demonstrate the role of early and intensive glycemic control in the prevention of micro and macrovascular disease in both type 1 and type 2 diabetes mellitus (DM). Hyperglycemia elicits several pathways related to the etiopathogenesis of cardiovascular disease (CVD), including the generation of advanced glycation end products (AGEs). In this review, we revisit the role played by AGEs in CVD based in clinical trials and experimental evidence. Mechanistic aspects concerning the recognition of AGEs by the advanced glycosylation end product-specific receptor (AGER) and its counterpart, the dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST) and soluble AGER are discussed. A special focus is offered to the AGE-elicited pathways that promote cholesterol accumulation in the arterial wall by enhanced oxidative stress, inflammation, endoplasmic reticulum stress and impairment in the reverse cholesterol transport (RCT).
Collapse
|
73
|
Bezhaeva T, Karper J, Quax PHA, de Vries MR. The Intriguing Role of TLR Accessory Molecules in Cardiovascular Health and Disease. Front Cardiovasc Med 2022; 9:820962. [PMID: 35237675 PMCID: PMC8884272 DOI: 10.3389/fcvm.2022.820962] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Activation of Toll like receptors (TLR) plays an important role in cardiovascular disease development, progression and outcomes. Complex TLR mediated signaling affects vascular and cardiac function including tissue remodeling and repair. Being central components of both innate and adaptive arms of the immune system, TLRs interact as pattern recognition receptors with a series of exogenous ligands and endogenous molecules or so-called danger associated molecular patterns (DAMPs) that are released upon tissue injury and cellular stress. Besides immune cells, a number of structural cells within the cardiovascular system, including endothelial cells, smooth muscle cells, fibroblasts and cardiac myocytes express TLRs and are able to release or sense DAMPs. Local activation of TLR-mediated signaling cascade induces cardiovascular tissue repair but in a presence of constant stimuli can overshoot and cause chronic inflammation and tissue damage. TLR accessory molecules are essential in guiding and dampening these responses toward an adequate reaction. Furthermore, accessory molecules assure specific and exclusive TLR-mediated signal transduction for distinct cells and pathways involved in the pathogenesis of cardiovascular diseases. Although much has been learned about TLRs activation in cardiovascular remodeling, the exact role of TLR accessory molecules is not entirely understood. Deeper understanding of the role of TLR accessory molecules in cardiovascular system may open therapeutic avenues aiming at manipulation of inflammatory response in cardiovascular disease. The present review outlines accessory molecules for membrane TLRs that are involved in cardiovascular disease progression. We first summarize the up-to-date knowledge on TLR signaling focusing on membrane TLRs and their ligands that play a key role in cardiovascular system. We then survey the current evidence of the contribution of TLRs accessory molecules in vascular and cardiac remodeling including myocardial infarction, heart failure, stroke, atherosclerosis, vein graft disease and arterio-venous fistula failure.
Collapse
Affiliation(s)
- Taisiya Bezhaeva
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Jacco Karper
- Department of Cardiology, Wilhelmina Hospital Assen, Assen, Netherlands
| | - Paul H. A. Quax
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Margreet R. de Vries
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Margreet R. de Vries
| |
Collapse
|
74
|
Qian J, Zhuang F, Chen Y, Fan X, Wang J, Wang Z, Wang Y, Xu M, Samorodov AV, Pavlov VN, Liang G. Myeloid differential protein-2 inhibition improves diabetic cardiomyopathy via p38MAPK inhibition and AMPK pathway activation. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166369. [PMID: 35176461 DOI: 10.1016/j.bbadis.2022.166369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
|
75
|
Zhang QY, Xu SJ, Qian JC, Yang LB, Chen PQ, Wang Y, Hu X, Zhang YL, Luo W, Liang G. Pharmacological inhibition of MyD88 suppresses inflammation in tubular epithelial cells and prevents diabetic nephropathy in experimental mice. Acta Pharmacol Sin 2022; 43:354-366. [PMID: 34552217 PMCID: PMC8792016 DOI: 10.1038/s41401-021-00766-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/12/2021] [Indexed: 02/03/2023]
Abstract
Emerging evidence shows that chronic inflammation mediated by toll-like receptors (TLRs) contributes to diabetic nephropathy. Myeloid differentiation primary-response protein-88 (MyD88) is an essential adapter protein of all TLRs except TLR3 in innate immunity. It is unclear whether MyD88 could be a therapeutic target for diabetic nephropathy. Here, we used a new small-molecule MyD88 inhibitor, LM8, to examine the pharmacological inhibition of MyD88 in protecting kidneys from inflammatory injury in diabetes. We showed that MyD88 was significantly activated in the kidney of STZ-induced type 1 diabetic mice in tubular epithelial cells as well as in high glucose-treated rat tubular epithelial cells NRK-52E. In cultured tubular epithelial cells, we show that LM8 (2.5-10 μM) or MyD88 siRNA attenuated high-concentration glucose-induced inflammatory and fibrogenic responses through inhibition of MyD88-TLR4 interaction and downstream NF-κB activation. Treatment with LM8 (5, 10 mg/kg, i.g.) significantly reduced renal inflammation and fibrosis and preserved renal function in both type 1 and type 2 diabetic mice. These renoprotective effects were associated with reduced MyD88-TLR4 complex formation, suppressed NF-κB signaling, and prevention of inflammatory factor expression. Collectively, our results show that hyperglycemia activates MyD88 signaling cascade to induce renal inflammation, fibrosis, and dysfunction. Pharmacological inhibition of MyD88 may be a therapeutic approach to mitigate diabetic nephropathy and the inhibitor LM8 could be a potential candidate for such therapy.
Collapse
Affiliation(s)
- Qiu-yan Zhang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.268099.c0000 0001 0348 3990Medical Research Center, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035 China
| | - Su-jing Xu
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.268099.c0000 0001 0348 3990School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027 China
| | - Jian-chang Qian
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Li-bin Yang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Peng-qin Chen
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Yi Wang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Xiang Hu
- grid.268099.c0000 0001 0348 3990Department of Endocrinology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035 China
| | - Ya-li Zhang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Wu Luo
- grid.268099.c0000 0001 0348 3990Medical Research Center, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035 China
| | - Guang Liang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.506977.a0000 0004 1757 7957School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399 China ,grid.410726.60000 0004 1797 8419Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001 China
| |
Collapse
|
76
|
Wang X, Chen X, Zhou W, Men H, Bao T, Sun Y, Wang Q, Tan Y, Keller BB, Tong Q, Zheng Y, Cai L. Ferroptosis is essential for diabetic cardiomyopathy and is prevented by sulforaphane via AMPK/NRF2 pathways. Acta Pharm Sin B 2022; 12:708-722. [PMID: 35256941 PMCID: PMC8897044 DOI: 10.1016/j.apsb.2021.10.005] [Citation(s) in RCA: 302] [Impact Index Per Article: 100.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/21/2021] [Accepted: 09/14/2021] [Indexed: 01/11/2023] Open
Abstract
Herein, we define the role of ferroptosis in the pathogenesis of diabetic cardiomyopathy (DCM) by examining the expression of key regulators of ferroptosis in mice with DCM and a new ex vivo DCM model. Advanced glycation end-products (AGEs), an important pathogenic factor of DCM, were found to induce ferroptosis in engineered cardiac tissues (ECTs), as reflected through increased levels of Ptgs2 and lipid peroxides and decreased ferritin and SLC7A11 levels. Typical morphological changes of ferroptosis in cardiomyocytes were observed using transmission electron microscopy. Inhibition of ferroptosis with ferrostatin-1 and deferoxamine prevented AGE-induced ECT remodeling and dysfunction. Ferroptosis was also evidenced in the heart of type 2 diabetic mice with DCM. Inhibition of ferroptosis by liproxstatin-1 prevented the development of diastolic dysfunction at 3 months after the onset of diabetes. Nuclear factor erythroid 2-related factor 2 (NRF2) activated by sulforaphane inhibited cardiac cell ferroptosis in both AGE-treated ECTs and hearts of DCM mice by upregulating ferritin and SLC7A11 levels. The protective effect of sulforaphane on ferroptosis was AMP-activated protein kinase (AMPK)-dependent. These findings suggest that ferroptosis plays an essential role in the pathogenesis of DCM; sulforaphane prevents ferroptosis and associated pathogenesis via AMPK-mediated NRF2 activation. This suggests a feasible therapeutic approach with sulforaphane to clinically prevent ferroptosis and DCM.
Collapse
Affiliation(s)
- Xiang Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Xinxin Chen
- Department of Burn Surgery, First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Wenqian Zhou
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Hongbo Men
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Terigen Bao
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Yike Sun
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Quanwei Wang
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bradley B. Keller
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Cincinnati Children's Heart Institute, Greater Louisville and Western Kentucky Practice, Louisville, KY 40202, USA
| | - Qian Tong
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Yang Zheng
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun 130021, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
77
|
Muñoz-Córdova F, Hernández-Fuentes C, Lopez-Crisosto C, Troncoso MF, Calle X, Guerrero-Moncayo A, Gabrielli L, Chiong M, Castro PF, Lavandero S. Novel Insights Into the Pathogenesis of Diabetic Cardiomyopathy and Pharmacological Strategies. Front Cardiovasc Med 2022; 8:707336. [PMID: 35004869 PMCID: PMC8734937 DOI: 10.3389/fcvm.2021.707336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe complication of diabetes developed mainly in poorly controlled patients. In DCM, several clinical manifestations as well as cellular and molecular mechanisms contribute to its phenotype. The production of reactive oxygen species (ROS), chronic low-grade inflammation, mitochondrial dysfunction, autophagic flux inhibition, altered metabolism, dysfunctional insulin signaling, cardiomyocyte hypertrophy, cardiac fibrosis, and increased myocardial cell death are described as the cardinal features involved in the genesis and development of DCM. However, many of these features can be associated with broader cellular processes such as inflammatory signaling, mitochondrial alterations, and autophagic flux inhibition. In this review, these mechanisms are critically discussed, highlighting the latest evidence and their contribution to the pathogenesis of DCM and their potential as pharmacological targets.
Collapse
Affiliation(s)
- Felipe Muñoz-Córdova
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Carolina Hernández-Fuentes
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Camila Lopez-Crisosto
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile
| | - Mayarling F Troncoso
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ximena Calle
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Alejandra Guerrero-Moncayo
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Luigi Gabrielli
- Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile
| | - Mario Chiong
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Pablo F Castro
- Division of Cardiovascular Diseases, Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), Pontifical Catholic University of Chile, Santiago, Chile.,Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.,Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), University of Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
78
|
Huang K, Luo X, Zhong Y, Deng L, Feng J. New insights into the role of melatonin in diabetic cardiomyopathy. Pharmacol Res Perspect 2022; 10:e00904. [PMID: 35005848 PMCID: PMC8929360 DOI: 10.1002/prp2.904] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetic cardiovascular complications and impaired cardiac function are considered to be the main causes of death in diabetic patients worldwide, especially patients with type 2 diabetes mellitus (T2DM). An increasing number of studies have shown that melatonin, as the main product secreted by the pineal gland, plays a vital role in the occurrence and development of diabetes. Melatonin improves myocardial cell metabolism, reduces vascular endothelial cell death, reverses microcirculation disorders, reduces myocardial fibrosis, reduces oxidative and endoplasmic reticulum stress, regulates cell autophagy and apoptosis, and improves mitochondrial function, all of which are the characteristics of diabetic cardiomyopathy (DCM). This review focuses on the role of melatonin in DCM. We also discuss new molecular findings that might facilitate a better understanding of the underlying mechanism. Finally, we propose potential new therapeutic strategies for patients with T2DM.
Collapse
Affiliation(s)
- Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xianling Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
79
|
Wan L, Bai X, Zhou Q, Chen C, Wang H, Liu T, Xue J, Wei C, Xie L. The advanced glycation end-products (AGEs)/ROS/NLRP3 inflammasome axis contributes to delayed diabetic corneal wound healing and nerve regeneration. Int J Biol Sci 2022; 18:809-825. [PMID: 35002527 PMCID: PMC8741862 DOI: 10.7150/ijbs.63219] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/27/2021] [Indexed: 12/31/2022] Open
Abstract
Diabetic keratopathy (DK) is an important diabetic complication at the ocular surface. Chronic low-grade inflammation mediated by the NLRP3 inflammasome promotes pathogenesis of diabetes and its complications. However, the effect of the NLRP3 inflammasome on DK pathogenesis remains elusive. Wild-type (WT) and Nlrp3 knockout (KO) C57 mice were used to establish a type I diabetes model by intraperitoneal injection of streptozotocin. The effect of the NLRP3 inflammasome on diabetic corneal wound healing and never regeneration was examined by a corneal epithelial abrasion model. Western blot, immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA) and pharmacological treatment were performed to investigate the regulatory mechanism of advanced glycation end products (AGEs) on NLRP3 inflammasome activation and corneal wound healing in vivo. The cultured mouse corneal epithelial cells (TKE2) were used to evaluate the effect and mechanism of AGEs on NLRP3 inflammasome activation in vitro. We revealed that NLRP3 inflammasome-mediated inflammation and pyroptosis contributed to DK pathogenesis. Under physiological conditions, the NLRP3 inflammasome was required for corneal wound healing and nerve regeneration. However, under a diabetic scenario, sustained activation of the NLRP3 inflammasome resulted in postponed corneal wound healing and impaired nerve regeneration. Mechanistically, the accumulated AGEs promoted hyperactivation of the NLRP3 inflammasome through ROS production. Moreover, genetically and pharmacologically blocking the AGEs/ROS/NLRP3 inflammasome axis significantly expedited diabetic corneal epithelial wound closure and nerve regeneration. Our results revealed that AGEs-induced hyperactivation of the NLRP3 inflammasome resulted in delayed diabetic corneal wound healing and impaired nerve regeneration, which further highlighted the NLRP3 inflammasome as a promising target for DK treatment.
Collapse
Affiliation(s)
- Luqin Wan
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Xiaofei Bai
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Chen Chen
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Huifeng Wang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Ting Liu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Junfa Xue
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, China
| |
Collapse
|
80
|
Zhou Q, Yang L, Wang Q, Li Y, Wei C, Xie L. Mechanistic investigations of diabetic ocular surface diseases. Front Endocrinol (Lausanne) 2022; 13:1079541. [PMID: 36589805 PMCID: PMC9800783 DOI: 10.3389/fendo.2022.1079541] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
With the global prevalence of diabetes mellitus over recent decades, more patients suffered from various diabetic complications, including diabetic ocular surface diseases that may seriously affect the quality of life and even vision sight. The major diabetic ocular surface diseases include diabetic keratopathy and dry eye. Diabetic keratopathy is characterized with the delayed corneal epithelial wound healing, reduced corneal nerve density, decreased corneal sensation and feeling of burning or dryness. Diabetic dry eye is manifested as the reduction of tear secretion accompanied with the ocular discomfort. The early clinical symptoms include dry eye and corneal nerve degeneration, suggesting the early diagnosis should be focused on the examination of confocal microscopy and dry eye symptoms. The pathogenesis of diabetic keratopathy involves the accumulation of advanced glycation end-products, impaired neurotrophic innervations and limbal stem cell function, and dysregulated growth factor signaling, and inflammation alterations. Diabetic dry eye may be associated with the abnormal mitochondrial metabolism of lacrimal gland caused by the overactivation of sympathetic nervous system. Considering the important roles of the dense innervations in the homeostatic maintenance of cornea and lacrimal gland, further studies on the neuroepithelial and neuroimmune interactions will reveal the predominant pathogenic mechanisms and develop the targeting intervention strategies of diabetic ocular surface complications.
Collapse
Affiliation(s)
- Qingjun Zhou
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Lingling Yang
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Ya Li
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- *Correspondence: Lixin Xie,
| |
Collapse
|
81
|
Zhang YL, Zhang WX, Yan JQ, Tang YL, Jia WJ, Xu ZW, Xu MJ, Chattipakorn N, Wang Y, Feng JP, Liu ZG, Liang G. Chalcone derivatives ameliorate lipopolysaccharide-induced acute lung injury and inflammation by targeting MD2. Acta Pharmacol Sin 2022; 43:76-85. [PMID: 34480112 PMCID: PMC8724327 DOI: 10.1038/s41401-021-00764-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/10/2021] [Indexed: 01/03/2023]
Abstract
Acute lung injury (ALI) and its severe form acute respiratory distress syndrome (ARDS) are known as the common causes of respiratory failure in critically ill patients. Myeloid differentiation 2 (MD2), a co-receptor of toll like receptor 4 (TLR4), plays an important role in LPS-induced ALI in mice. Since MD2 inhibition by pharmacological inhibitors or gene knockout significantly attenuates ALI in animal models, MD2 has become an attractive target for the treatment of ALI. In this study we identified two chalcone-derived compounds, 7w and 7x, as new MD2 inhibitors, and investigated the therapeutic effects of 7x and 7w in LPS-induced ALI mouse model. In molecular docking analysis we found that 7w and 7x, formed pi-pi stacking interactions with Phe151 residue of the MD2 protein. The direct binding was confirmed by surface plasmon resonance analysis (with KD value of 96.2 and 31.2 μM, respectively) and by bis-ANS displacement assay. 7w and 7x (2.5, 10 μM) also dose-dependently inhibited the interaction between lipopolysaccharide (LPS) and rhMD2 and LPS-MD2-TLR4 complex formation. In mouse peritoneal macrophages, 7w and 7x (1.25-10 μM) dose-dependently inhibited LPS-induced inflammatory responses, MAPKs (JNK, ERK and P38) phosphorylation as well as NF-κB activation. Finally, oral administration of 7w or 7x (10 mg ·kg-1 per day, for 7 days prior LPS challenge) in ALI mouse model significantly alleviated LPS-induced lung injury, pulmonary edema, lung permeability, inflammatory cells infiltration, inflammatory cytokines expression and MD2/TLR4 complex formation. In summary, we identify 7w and 7x as new MD2 inhibitors to inhibit inflammatory response both in vitro and in vivo, proving the therapeutic potential of 7w and 7x for ALI and inflammatory diseases.
Collapse
Affiliation(s)
- Ya-li Zhang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.440761.00000 0000 9030 0162Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005 China ,grid.506977.a0000 0004 1757 7957School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399 China
| | - Wen-xin Zhang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.268099.c0000 0001 0348 3990Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, 311800 China
| | - Jue-qian Yan
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Ye-lin Tang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Wen-jing Jia
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.268099.c0000 0001 0348 3990Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, 311800 China
| | - Zheng-wei Xu
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Ming-jiang Xu
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Nipon Chattipakorn
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Yi Wang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Jian-peng Feng
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China
| | - Zhi-guo Liu
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.268099.c0000 0001 0348 3990Zhuji Biomedicine Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, 311800 China
| | - Guang Liang
- grid.268099.c0000 0001 0348 3990Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 China ,grid.506977.a0000 0004 1757 7957School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399 China ,grid.410726.60000 0004 1797 8419Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001 China
| |
Collapse
|
82
|
Peng L, Zhu M, Huo S, Shi W, Jiang T, Peng D, Wang M, Jiang Y, Guo J, Men L, Huang B, Wang Q, Lv J, Lin L, Li S. Myocardial protection of S-nitroso-L-cysteine in diabetic cardiomyopathy mice. Front Endocrinol (Lausanne) 2022; 13:1011383. [PMID: 36313766 PMCID: PMC9602402 DOI: 10.3389/fendo.2022.1011383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/27/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe complication of diabetes mellitus that is characterized by aberrant myocardial structure and function and is the primary cause of heart failure and death in diabetic patients. Endothelial dysfunction plays an essential role in diabetes and is associated with an increased risk of cardiovascular events, but its role in DCM is unclear. Previously, we showed that S-nitroso-L-cysteine(CSNO), an endogenous S-nitrosothiol derived from eNOS, inhibited the activity of protein tyrosine phosphatase 1B (PTP1B), a critical negative modulator of insulin signaling. In this study, we reported that CSNO treatment induced cellular insulin-dependent and insulin-independent glucose uptake. In addition, CSNO activated insulin signaling pathway and promoted GLUT4 membrane translocation. CSNO protected cardiomyocytes against high glucose-induced injury by ameliorating excessive autophagy activation, mitochondrial impairment and oxidative stress. Furthermore, nebulized CSNO improved cardiac function and myocardial fibrosis in diabetic mice. These results suggested a potential site for endothelial modulation of insulin sensitivity and energy metabolism in the development of DCM. Data from these studies will not only help us understand the mechanisms of DCM, but also provide new therapeutic options for treatment.
Collapse
Affiliation(s)
- Lulu Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengying Zhu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengqi Huo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Shi
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dewei Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Moran Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Guo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lintong Men
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingyu Huang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Sheng Li, ;
| |
Collapse
|
83
|
Peng ML, Fu Y, Wu CW, Zhang Y, Ren H, Zhou SS. Signaling Pathways Related to Oxidative Stress in Diabetic Cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:907757. [PMID: 35784531 PMCID: PMC9240190 DOI: 10.3389/fendo.2022.907757] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a chronic metabolic disease that is increasing in prevalence and causes many complications. Diabetic cardiomyopathy (DCM) is a complication of diabetes that is associated with high mortality, but it is not well defined. Nevertheless, it is generally accepted that DCM refers to a clinical disease that occurs in patients with diabetes and involves ventricular dysfunction, in the absence of other cardiovascular diseases, such as coronary atherosclerotic heart disease, hypertension, or valvular heart disease. However, it is currently uncertain whether the pathogenesis of DCM is directly attributable to metabolic dysfunction or secondary to diabetic microangiopathy. Oxidative stress (OS) is considered to be a key component of its pathogenesis. The production of reactive oxygen species (ROS) in cardiomyocytes is a vicious circle, resulting in further production of ROS, mitochondrial DNA damage, lipid peroxidation, and the post-translational modification of proteins, as well as inflammation, cardiac hypertrophy and fibrosis, ultimately leading to cell death and cardiac dysfunction. ROS have been shown to affect various signaling pathways involved in the development of DCM. For instance, OS causes metabolic disorders by affecting the regulation of PPARα, AMPK/mTOR, and SIRT3/FOXO3a. Furthermore, OS participates in inflammation mediated by the NF-κB pathway, NLRP3 inflammasome, and the TLR4 pathway. OS also promotes TGF-β-, Rho-ROCK-, and Notch-mediated cardiac remodeling, and is involved in the regulation of calcium homeostasis, which impairs ATP production and causes ROS overproduction. In this review, we summarize the signaling pathways that link OS to DCM, with the intention of identifying appropriate targets and new antioxidant therapies for DCM.
Collapse
Affiliation(s)
- Meng-ling Peng
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Yu Fu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chu-wen Wu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Hang Ren
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Shan-shan Zhou
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Shan-shan Zhou,
| |
Collapse
|
84
|
Astragaloside IV protects diabetic cardiomyopathy against inflammation and apoptosis via regulating TLR4/MyD88/NF-κB signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
85
|
Zhu W, Sahar NE, Javaid HMA, Pak ES, Liang G, Wang Y, Ha H, Huh JY. Exercise-Induced Irisin Decreases Inflammation and Improves NAFLD by Competitive Binding with MD2. Cells 2021; 10:3306. [PMID: 34943814 PMCID: PMC8699279 DOI: 10.3390/cells10123306] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global clinical problem. The MD2-TLR4 pathway exacerbates NAFLD progression by promoting inflammation. Long-term exercise is considered to improve NAFLD but the underlying mechanism is still unclear. In this study, we examined the protective effect and molecular mechanism of exercise on high-fat diet (HFD)-induced liver injury. In an HFD-induced NAFLD mouse model, exercise training significantly decreased hepatic steatosis and fibrosis. Interestingly, exercise training blocked the binding of MD2-TLR4 and decreased the downstream inflammatory response. Irisin is a myokine that is highly expressed in response to exercise and exerts anti-inflammatory effects. We found that circulating irisin levels and muscle irisin expression were significantly increased in exercised mice, suggesting that irisin could mediate the effect of exercise on NAFLD. In vitro studies showed that irisin improved lipid metabolism, fibrosis, and inflammation in palmitic acid (PA)-stimulated AML12 cells. Moreover, binding assay results showed that irisin disturbed MD2-TLR4 complex formation by directly binding with MD2 but not TLR4, and interfered with the recognition of stimuli such as PA and lipopolysaccharide with MD2. Our study provides novel evidence that exercise-induced irisin inhibits inflammation via competitive binding with MD2 to improve NAFLD. Thus, irisin could be considered a potential therapy for NAFLD.
Collapse
Affiliation(s)
- Weiwei Zhu
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (W.Z.); (N.E.S.); (H.M.A.J.)
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (G.L.); (Y.W.)
| | - Namood E Sahar
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (W.Z.); (N.E.S.); (H.M.A.J.)
| | | | - Eun Seon Pak
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (E.S.P.); (H.H.)
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (G.L.); (Y.W.)
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (G.L.); (Y.W.)
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (E.S.P.); (H.H.)
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (W.Z.); (N.E.S.); (H.M.A.J.)
| |
Collapse
|
86
|
Inhibition of myeloid differentiation factor 2 attenuates cardiometabolic impairments via reducing cardiac mitochondrial dysfunction, inflammation, apoptosis and ferroptosis in prediabetic rats. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166301. [PMID: 34748903 DOI: 10.1016/j.bbadis.2021.166301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022]
Abstract
Systemic inflammation is a key mediator of left ventricular dysfunction (LV) in prediabetes via the activation of myeloid differentiation factor 2 (MD2)/toll-like receptor 4 complex. The MD2 inhibitor L6H21 effectively reduced systemic and cardiac inflammation in obese mice. However, its effects on cardiac function and regulated cell death pathways in the heart in prediabetes are still unknown. The prediabetic rats were divided into 3 subgroups to receive vehicle, L6H21 (10, 20, 40 mg/kg) or metformin (300 mg/kg) for 1, 2 and 4 weeks. Then, metabolic parameters, cardiac sympathovagal balance, LV function, cardiac mitochondrial function, oxidative stress, inflammation, apoptosis, necroptosis, and ferroptosis were determined. All prediabetic rats exhibited cardiac sympathovagal imbalance, LV dysfunction, and cardiac mitochondrial dysfunction. All doses of L6H21 treatment for 2- and 4-weeks attenuated insulin resistance. L6H21 at 40 mg/kg attenuated cardiac autonomic imbalance and LV dysfunction after 1 week of treatment. Both 10 and 20 mg/kg of L6H21 required longer treatment duration to show these benefits. Mechanistically, all doses of L6H21 reduced cardiac mitochondrial dysfunction after 1 week of treatment, resulting in alleviated oxidative stress and inflammation. L6H21 also effectively suppressed cardiac apoptosis and ferroptosis, but it did not affect necroptosis in prediabetic rats. L6H21 provided the cardioprotective efficacy in dose- and time-dependent manners in prediabetic rats via reduction in apoptosis and ferroptosis.
Collapse
|
87
|
Zuo W, Zhao J, Zhang J, Fang Z, Deng J, Fan Z, Guo Y, Han J, Hou W, Dong H, Xu F, Xiong L. MD2 contributes to the pathogenesis of perioperative neurocognitive disorder via the regulation of α5GABA A receptors in aged mice. J Neuroinflammation 2021; 18:204. [PMID: 34530841 PMCID: PMC8444589 DOI: 10.1186/s12974-021-02246-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorder (PND) is a long-term postoperative complication in elderly surgical patients. The underlying mechanism of PND is unclear, and no effective therapies are currently available. It is believed that neuroinflammation plays an important role in triggering PND. The secreted glycoprotein myeloid differentiation factor 2 (MD2) functions as an activator of the Toll-like receptor 4 (TLR4) inflammatory pathway, and α5GABAA receptors (α5GABAARs) are known to play a key role in regulating inflammation-induced cognitive deficits. Thus, in this study, we aimed to investigate the role of MD2 in PND and determine whether α5GABAARs are involved in the function of MD2. METHODS Eighteen-month-old C57BL/6J mice were subjected to laparotomy under isoflurane anesthesia to induce PND. The Barnes maze was used to assess spatial reference learning and memory, and the expression of hippocampal MD2 was assayed by western blotting. MD2 expression was downregulated by bilateral injection of AAV-shMD2 into the hippocampus or tail vein injection of the synthetic MD2 degrading peptide Tat-CIRP-CMA (TCM) to evaluate the effect of MD2. Primary cultured neurons from brain tissue block containing cortices and hippocampus were treated with Tat-CIRP-CMA to investigate whether downregulating MD2 expression affected the expression of α5GABAARs. Electrophysiology was employed to measure tonic currents. For α5GABAARs intervention experiments, L-655,708 and L-838,417 were used to inhibit or activate α5GABAARs, respectively. RESULTS Surgery under inhaled isoflurane anesthesia induced cognitive impairments and elevated the expression of MD2 in the hippocampus. Downregulation of MD2 expression by AAV-shMD2 or Tat-CIRP-CMA improved the spatial reference learning and memory in animals subjected to anesthesia and surgery. Furthermore, Tat-CIRP-CMA treatment decreased the expression of membrane α5GABAARs and tonic currents in CA1 pyramidal neurons in the hippocampus. Inhibition of α5GABAARs by L-655,708 alleviated cognitive impairments after anesthesia and surgery. More importantly, activation of α5GABAARs by L-838,417 abrogated the protective effects of Tat-CIRP-CMA against anesthesia and surgery-induced spatial reference learning and memory deficits. CONCLUSIONS MD2 contributes to the occurrence of PND by regulating α5GABAARs in aged mice, and Tat-CIRP-CMA is a promising neuroprotectant against PND.
Collapse
Affiliation(s)
- Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jinming Zhang
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, 710062, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ze Fan
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yaru Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, 710062, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Feifei Xu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China. .,Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence Affiliated to Tongji University School of Medicine, Shanghai, 200434, China.
| |
Collapse
|
88
|
Krasnodębski M, Grąt K, Morawski M, Borkowski J, Krawczyk P, Zhylko A, Skalski M, Kalinowski P, Zieniewicz K, Grąt M. Skin autofluorescence as a novel predictor of acute kidney injury after liver resection. World J Surg Oncol 2021; 19:276. [PMID: 34526025 PMCID: PMC8444415 DOI: 10.1186/s12957-021-02394-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/03/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Skin autofluorescence (SAF) reflects accumulation of advanced glycation end-products (AGEs). The aim of this study was to evaluate predictive usefulness of SAF measurement in prediction of acute kidney injury (AKI) after liver resection. METHODS This prospective observational study included 130 patients undergoing liver resection. The primary outcome measure was AKI. SAF was measured preoperatively and expressed in arbitrary units (AU). RESULTS AKI was observed in 32 of 130 patients (24.6%). SAF independently predicted AKI (p = 0.047), along with extent of resection (p = 0.019) and operative time (p = 0.046). Optimal cut-off for SAF in prediction of AKI was 2.7 AU (area under the curve [AUC] 0.611), with AKI rates of 38.7% and 20.2% in patients with high and low SAF, respectively (p = 0.037). Score based on 3 independent predictors (SAF, extent of resection, and operative time) well stratified the risk of AKI (AUC 0.756), with positive and negative predictive values of 59.3% and 84.0%, respectively. In particular, SAF predicted AKI in patients undergoing major and prolonged resections (p = 0.010, AUC 0.733) with positive and negative predictive values of 81.8%, and 62.5%, respectively. CONCLUSIONS AGEs accumulation negatively affects renal function in patients undergoing liver resection. SAF measurement may be used to predict AKI after liver resection, particularly in high-risk patients.
Collapse
Affiliation(s)
- Maciej Krasnodębski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland.
| | - Karolina Grąt
- Second Department of Clinical Radiology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Morawski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Jan Borkowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Krawczyk
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Andriy Zhylko
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Michał Skalski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Kalinowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Zieniewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
89
|
Qi Y, Fang Q, Li Q, Ding H, Shu Q, Hu Y, Xin W, Fang L. MD2 blockage prevents the migration and invasion of hepatocellular carcinoma cells via inhibition of the EGFR signaling pathway. J Gastrointest Oncol 2021; 12:1873-1883. [PMID: 34532135 PMCID: PMC8421902 DOI: 10.21037/jgo-21-362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The toll-like receptor (TLR) is an emerging signaling pathway in tumor invasion and metastasis. The activation of TLRs requires specific accessory proteins, such as the small secreted glycoprotein myeloid differentiation protein 2 (MD2), which contributes to ligand responsiveness. However, the role of MD2 in tumorigenesis and metastasis has rarely been reported. This study aimed to investigate the effects and underlying mechanisms of MD2 on the proliferation, migration, and invasion of hepatocellular carcinoma (HCC). METHODS Cell counting kit 8 (CCK8), cell colony formation, wound healing, and transwell assays were conducted to determine cell viability, proliferation, migration, and invasion, respectively. Quantitative real-time PCR (qRT-PCR) was performed to assess the expression of MD2 in HCC cell lines and human normal liver cell lines as well as the silencing efficiency of MD2 blockage. Western blot and qRT-PCR assays were performed to detect the protein and mRNA expression levels of epithelial mesenchymal transformation (EMT) markers and epidermal growth factor receptor (EGFR) signaling molecules. RESULTS MD2 was highly expressed in HCC tissues and cell lines. High expression of MD2 was associated with poor prognosis of HCC patients. In addition, MD2 silencing slightly inhibited the proliferation of HepG2 and HCCLM3, and significantly suppressed cell migration and invasion. Furthermore, MD2 blockage could distinctly prevent the EMT process by increasing the protein and mRNA levels of E-cadherin and Occludin, and decreasing the levels of Vimentin, N-cadherin, and Snail. Finally, the phosphorylation level of EGFR as well as its downstream molecular Src, Akt, I-κBα, and p65 were downregulated in HCC cells with MD2 silencing. CONCLUSIONS Our findings suggest that high expression of MD2 may affect the EMT, migration, and invasion via modulation of the EGFR pathway in HCC cells.
Collapse
Affiliation(s)
- Yajun Qi
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qilu Fang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qinglin Li
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Department of Comprehensive Medical Oncology, Key Laboratory of Head and Neck Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haiying Ding
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Qi Shu
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yan Hu
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wenxiu Xin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Department of Comprehensive Medical Oncology, Key Laboratory of Head and Neck Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Luo Fang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
90
|
Ju W, Liu K, Ouyang S, Liu Z, He F, Wu J. Changes in N6-Methyladenosine Modification Modulate Diabetic Cardiomyopathy by Reducing Myocardial Fibrosis and Myocyte Hypertrophy. Front Cell Dev Biol 2021; 9:702579. [PMID: 34368154 PMCID: PMC8334868 DOI: 10.3389/fcell.2021.702579] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/21/2021] [Indexed: 01/23/2023] Open
Abstract
In this study, we aimed to systematically profile global RNA N6-methyladenosine (m6A) modification patterns in a mouse model of diabetic cardiomyopathy (DCM). Patterns of m6A in DCM and normal hearts were analyzed via m6A-specific methylated RNA immunoprecipitation followed by high-throughput sequencing (MeRIP-seq) and RNA sequencing (RNA-seq). m6A-related mRNAs were validated by quantitative real-time PCR analysis of input and m6A immunoprecipitated RNA samples from DCM and normal hearts. A total of 973 new m6A peaks were detected in DCM samples and 984 differentially methylated sites were selected for further study, including 295 hypermethylated and 689 hypomethylated m6A sites (fold change (FC) > 1.5, P < 0.05). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway analyses indicated that unique m6A-modified transcripts in DCM were closely linked to cardiac fibrosis, myocardial hypertrophy, and myocardial energy metabolism. Total m6A levels were higher in DCM, while levels of the fat mass and obesity-associated (FTO) protein were downregulated. Overexpression of FTO in DCM model mice improved cardiac function by reducing myocardial fibrosis and myocyte hypertrophy. Overall, m6A modification patterns were altered in DCM, and modification of epitranscriptomic processes, such as m6A, is a potentially interesting therapeutic approach.
Collapse
Affiliation(s)
- Wenhao Ju
- Graduate School, Peking Union Medical College, Beijing, China.,Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China
| | - Kai Liu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China.,Department of Biochemistry & Immunology, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Shengrong Ouyang
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China
| | - Zhuo Liu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China
| | - Feng He
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China
| | - Jianxin Wu
- Graduate School, Peking Union Medical College, Beijing, China.,Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.,Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China.,Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
91
|
Kaur N, Guan Y, Raja R, Ruiz-Velasco A, Liu W. Mechanisms and Therapeutic Prospects of Diabetic Cardiomyopathy Through the Inflammatory Response. Front Physiol 2021; 12:694864. [PMID: 34234695 PMCID: PMC8257042 DOI: 10.3389/fphys.2021.694864] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
The incidence of heart failure (HF) continues to increase rapidly in patients with diabetes. It is marked by myocardial remodeling, including fibrosis, hypertrophy, and cell death, leading to diastolic dysfunction with or without systolic dysfunction. Diabetic cardiomyopathy (DCM) is a distinct myocardial disease in the absence of coronary artery disease. DCM is partially induced by chronic systemic inflammation, underpinned by a hostile environment due to hyperglycemia, hyperlipidemia, hyperinsulinemia, and insulin resistance. The detrimental role of leukocytes, cytokines, and chemokines is evident in the diabetic heart, yet the precise role of inflammation as a cause or consequence of DCM remains incompletely understood. Here, we provide a concise review of the inflammatory signaling mechanisms contributing to the clinical complications of diabetes-associated HF. Overall, the impact of inflammation on the onset and development of DCM suggests the potential benefits of targeting inflammatory cascades to prevent DCM. This review is tailored to outline the known effects of the current anti-diabetic drugs, anti-inflammatory therapies, and natural compounds on inflammation, which mitigate HF progression in diabetic populations.
Collapse
Affiliation(s)
| | | | | | | | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
92
|
Han B, Xu J, Shi X, Zheng Z, Shi F, Jiang F, Han J. DL-3-n-Butylphthalide Attenuates Myocardial Hypertrophy by Targeting Gasdermin D and Inhibiting Gasdermin D Mediated Inflammation. Front Pharmacol 2021; 12:688140. [PMID: 34168567 PMCID: PMC8217660 DOI: 10.3389/fphar.2021.688140] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/26/2021] [Indexed: 11/18/2022] Open
Abstract
Pressure overload leads to a hypertrophic milieu that produces deleterious cardiac dysfunction. Inflammation is a key pathophysiological mechanism underpinning myocardial hypertrophy. DL-3-n-butylphthalide (NBP), a neuroprotective agent, also has potent cardioprotective effects. In this study, the potential of NBP to antagonize myocardial hypertrophy was evaluated in C57BL/6 mice in vivo and in rat primary cardiomyocytes in vitro. In mice, NBP treatment reduced cardiac hypertrophy and dysfunction in a transverse aortic constriction (TAC)-induced pressure overload model. In angiotensin (Ang) II-challenged cardiomyocytes, NBP prevents cell size increases and inhibits gasdermin D (GSDMD)-mediated inflammation. Furthermore, overexpression of GSDMD-N reduced the protective effects of NBP against Ang II-induced changes. Using molecular docking and MD simulation, we found that the GSDMD-N protein may be a target of NBP. Our study shows that NBP attenuates myocardial hypertrophy by targeting GSDMD and inhibiting GSDMD-mediated inflammation.
Collapse
Affiliation(s)
- Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiajun Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaowen Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhanxiong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Fengjie Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Fenfen Jiang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
93
|
Wang X, Chen XX, Yu HT, Tan Y, Lin Q, Keller BB, Zheng Y, Cai L. Engineered cardiac tissues: a novel in vitro model to investigate the pathophysiology of mouse diabetic cardiomyopathy. Acta Pharmacol Sin 2021; 42:932-941. [PMID: 33037406 PMCID: PMC8149662 DOI: 10.1038/s41401-020-00538-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/13/2020] [Indexed: 01/12/2023]
Abstract
Rodent diabetic models, used to understand the pathophysiology of diabetic cardiomyopathy (DCM), remain several limitations. Engineered cardiac tissues (ECTs) have emerged as robust 3D in vitro models to investigate structure-function relationships as well as cardiac injury and repair. Advanced glycation end-products (AGEs), produced through glycation of proteins or lipids in response to hyperglycemia, are important pathogenic factor for the development of DCM. In the current study, we developed a murine-based ECT model to investigate cardiac injury produced by AGEs. We treated ECTs composed of neonatal murine cardiac cells with AGEs and observed AGE-related functional, cellular, and molecular alterations: (1) AGEs (150 µg/mL) did not cause acute cytotoxicity, which displayed as necrosis detected by medium LDH release or apoptosis detected by cleaved caspase 3 and TUNEL staining, but negatively impacted ECT function on treatment day 9; (2) AGEs treatment significantly increased the markers of fibrosis (TGF-β, α-SMA, Ctgf, Collagen I-α1, Collagen III-α1, and Fn1) and hypertrophy (Nppa and Myh7); (3) AGEs treatment significantly increased ECT oxidative stress markers (3-NT, 4-HNE, HO-1, CAT, and SOD2) and inflammation response markers (PAI-1, TNF-α, NF-κB, and ICAM-1); and (4) AGE-induced pathogenic responses were all attenuated by pre-application of AGE receptor antagonist FPS-ZM1 (20 µM) or the antioxidant glutathione precursor N-acetylcysteine (5 mM). Therefore, AGEs-treated murine ECTs recapitulate the key features of DCM's functional, cellular and molecular pathogenesis, and may serve as a robust in vitro model to investigate cellular structure-function relationships, signaling pathways relevant to DCM and pharmaceutical intervention strategies.
Collapse
Affiliation(s)
- Xiang Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, 40202, USA
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xin-Xin Chen
- Department of Burn Surgery, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Hai-Tao Yu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, 40202, USA
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Qian Lin
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, 40202, USA
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
- Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Cincinnati Children's Heart Institute, Greater Louisville and Western Kentucky Practice, Louisville, KY, 40202, USA
| | - Yang Zheng
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
94
|
Huang S, You S, Qian J, Dai C, Shen S, Wang J, Huang W, Liang G, Wu G. Myeloid differentiation 2 deficiency attenuates AngII-induced arterial vascular oxidative stress, inflammation, and remodeling. Aging (Albany NY) 2021; 13:4409-4427. [PMID: 33495414 PMCID: PMC7906178 DOI: 10.18632/aging.202402] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/15/2020] [Indexed: 11/25/2022]
Abstract
Vascular remodeling is a pertinent target for cardiovascular therapy. Vascular smooth muscle cell (VSMC) dysfunction plays a key role in vascular remodeling. Myeloid differentiation 2 (MD2), a cofactor of toll-like receptor 4 (TLR4), is involved in atherosclerotic progress and cardiac remodeling via activation of chronic inflammation. In this study, we explored the role of MD2 in vascular remodeling using an Ang II-induced mouse model and cultured human aortic VSMCs. MD2 deficiency suppressed Ang II-induced vascular fibrosis and phenotypic switching of VSMCs without affecting blood pressure in mice. Mechanistically, MD2 deficiency prevented Ang II-induced expression of inflammatory cytokines and oxidative stress in mice and cultured VSMCs. Furthermore, MD2 deficiency reversed Ang II-activated MAPK signaling and Ang II-downregulated SIRT1 expression. Taken together, MD2 plays a significant role in Ang II-induced vascular oxidative stress, inflammation, and remodeling, indicating that MD2 is a potential therapeutic target for the treatment of vascular remodeling-related cardiovascular diseases.
Collapse
Affiliation(s)
- Shushi Huang
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengban You
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinfu Qian
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengyi Dai
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Wang
- Department of Cardiology, Affiliated Dingli Institute and Wenzhou Central Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Cardiology, Affiliated Dingli Institute and Wenzhou Central Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
95
|
Xiao M, Tang Y, Wang S, Wang J, Wang J, Guo Y, Zhang J, Gu J. The Role of Fibroblast Growth Factor 21 in Diabetic Cardiovascular Complications and Related Epigenetic Mechanisms. Front Endocrinol (Lausanne) 2021; 12:598008. [PMID: 34349728 PMCID: PMC8326758 DOI: 10.3389/fendo.2021.598008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21), is an emerging metabolic regulator mediates multiple beneficial effects in the treatment of metabolic disorders and related complications. Recent studies showed that FGF21 acts as an important inhibitor in the onset and progression of cardiovascular complications of diabetes mellitus (DM). Furthermore, evidences discussed so far demonstrate that epigenetic modifications exert a crucial role in the initiation and development of DM-related cardiovascular complications. Thus, epigenetic modifications may involve in the function of FGF21 on DM-induced cardiovascular complications. Therefore, this review mainly interprets and delineates the recent advances of role of FGF21 in DM cardiovascular complications. Then, the possible changes of epigenetics related to the role of FGF21 on DM-induced cardiovascular complications are discussed. Thus, this article not only implies deeper understanding of the pathological mechanism of DM-related cardiovascular complications, but also provides the possible novel therapeutic strategy for DM-induced cardiovascular complications by targeting FGF21 and related epigenetic mechanism.
Collapse
Affiliation(s)
- Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People’s Hospital of Liaoning Province, Shenyang, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Junlian Gu,
| |
Collapse
|
96
|
Wang X, Antony V, Wang Y, Wu G, Liang G. Pattern recognition receptor-mediated inflammation in diabetic vascular complications. Med Res Rev 2020; 40:2466-2484. [PMID: 32648967 DOI: 10.1002/med.21711] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 01/03/2025]
Abstract
The innate immune system contains multiple classes of pattern recognition receptors (PRRs), which recognize pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) in the intracellular and extracellular space. Although PRRs are indispensable for the detection and clearance of invading pathogens, dysregulated PRR activation by extrinsic and intrinsic factors leads to inflammatory diseases. PRR-mediated inflammation has been shown to play a pivotal role in the pathogenesis of diabetic vascular complications (DVCs), which are the leading causes of morbidity and mortality in diabetic patients. Upon sensing hyperglycemia-generated DAMPs, PRRs activate intracellular signaling pathways leading to the production of proinflammatory cytokines and chemokines in various cells of the kidney, brain, eye, and heart. The resulting chronic, low-grade inflammation contributes to DVCs. In this review, we summarize the role of PRRs in DVCs including diabetic nephropathy, neuropathy, retinopathy, and cardiomyopathy. We propose that targeting PRRs and associated signaling pathways may be beneficial for the management of DVCs.
Collapse
Affiliation(s)
- Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Victor Antony
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhuji Biomedical Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhuji Biomedical Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang, China
| |
Collapse
|
97
|
Wang X, Wang Y, Antony V, Sun H, Liang G. Metabolism-Associated Molecular Patterns (MAMPs). Trends Endocrinol Metab 2020; 31:712-724. [PMID: 32807598 DOI: 10.1016/j.tem.2020.07.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/06/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Metabolic diseases pose a tremendous health threat in both developed and developing countries. The pathophysiology of metabolic diseases is complex but has been shown to be closely associated with sterile inflammation, which is initiated by various danger molecules derived from metabolic overload, such as oxidized low-density lipoproteins (OxLDLs), free fatty acids (FFAs), glucose, advanced glycation end products (AGEs), and cholesterol. These danger signals are sensed by pattern recognition receptors (PRRs) to activate proinflammatory signaling pathways and promote the release of proinflammatory mediators, leading to chronic low-grade inflammation. Although these harmful metabolic stimuli are generally regarded as damage-associated molecular patterns (DAMPs), a more specific definition and accurate classification for these DAMPs is still missing. In this opinion, we classify the harmful metabolic stimuli that can incite inflammatory responses and tissue damage via instigating PRRs as metabolism-associated molecular patterns (MAMPs), and we summarize their roles in metaflammation-mediated metabolic diseases.
Collapse
Affiliation(s)
- Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Victor Antony
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhuji Biomedical Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang 311800, China.
| |
Collapse
|